1
|
Todhunter ME, Jubair S, Verma R, Saqe R, Shen K, Duffy B. Artificial intelligence and machine learning applications for cultured meat. Front Artif Intell 2024; 7:1424012. [PMID: 39381621 PMCID: PMC11460582 DOI: 10.3389/frai.2024.1424012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024] Open
Abstract
Cultured meat has the potential to provide a complementary meat industry with reduced environmental, ethical, and health impacts. However, major technological challenges remain which require time-and resource-intensive research and development efforts. Machine learning has the potential to accelerate cultured meat technology by streamlining experiments, predicting optimal results, and reducing experimentation time and resources. However, the use of machine learning in cultured meat is in its infancy. This review covers the work available to date on the use of machine learning in cultured meat and explores future possibilities. We address four major areas of cultured meat research and development: establishing cell lines, cell culture media design, microscopy and image analysis, and bioprocessing and food processing optimization. In addition, we have included a survey of datasets relevant to CM research. This review aims to provide the foundation necessary for both cultured meat and machine learning scientists to identify research opportunities at the intersection between cultured meat and machine learning.
Collapse
Affiliation(s)
| | - Sheikh Jubair
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Ruchika Verma
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Rikard Saqe
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Kevin Shen
- Department of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | | |
Collapse
|
2
|
Yu Y, Ouyang W, Huang Y, Huang H, Wang Z, Jia X, Huang Z, Lin R, Zhu Y, yalikun Y, Tan L, Li X, Zhao F, Chen Z, Li W, Liao J, Yao H, Long M. AI-Based multimodal Multi-tasks analysis reveals tumor molecular heterogeneity, predicts preoperative lymph node metastasis and prognosis in papillary thyroid carcinoma: A retrospective study. Int J Surg 2024; 111:01279778-990000000-01786. [PMID: 38990290 PMCID: PMC11745641 DOI: 10.1097/js9.0000000000001875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the predominant form of thyroid cancer globally, especially when lymph node metastasis (LNM) occurs. Molecular heterogeneity, driven by genetic alterations and tumor microenvironment components, contributes to the complexity of PTC. Understanding these complexities is essential for precise risk stratification and therapeutic decisions. METHODS This study involved a comprehensive analysis of 521 patients with PTC from our hospital and 499 patients from The Cancer Genome Atlas (TCGA). The real-world cohort 1 comprised 256 patients with stage I-III PTC. Tissues from 252 patients were analyzed by DNA-based next-generation sequencing, and tissues from four patients were analyzed by single-cell RNA sequencing (scRNA-seq). Additionally, 586 PTC pathological sections were collected from TCGA, and 275 PTC pathological sections were collected from the real-world cohort 2. A deep learning multimodal model was developed using matched histopathology images, genomic, transcriptomic, and immune cell data to predict LNM and disease-free survival (DFS). RESULTS This study included a total of 1,011 PTC patients, comprising 256 patients from cohort 1, 275 patients from cohort 2, and 499 patients from TCGA. In cohort 1, we categorized PTC into four molecular subtypes based on BRAF, RAS, RET, and other mutations. BRAF mutations were significantly associated with LNM and impacted DFS. ScRNA-seq identified distinct T cell subtypes and reduced B cell diversity in BRAF-mutated PTC with LNM. The study also explored cancer-associated fibroblasts and macrophages, highlighting their associations with LNM. The deep learning model was trained using 405 pathology slides and RNA sequences from 328 PTC patients and validated with 181 slides and RNA sequences from 140 PTC patients in the TCGA cohort. It achieved high accuracy, with an AUC of 0.86 in the training cohort, 0.84 in the validation cohort, and 0.83 in the real-world cohort 2. High-risk patients in the training cohort had significantly lower DFS rates (P<0.001). Model AUCs were 0.91 at 1 year, 0.93 at 3 years, and 0.87 at 5 years. In the validation cohort, high-risk patients also had lower DFS (P<0.001); the AUCs were 0.89, 0.87, and 0.80 at 1, 3, and 5 years. We utilized the GradCAM algorithm to generate heatmaps from pathology-based deep learning models, which visually highlighted high-risk tumor areas in PTC patients. This enhanced clinicians' understanding of the model's predictions and improved diagnostic accuracy, especially in cases with lymph node metastasis. CONCLUSION The AI-based analysis uncovered vital insights into PTC molecular heterogeneity, emphasizing BRAF mutations' impact. The integrated deep learning model shows promise in predicting metastasis, offering valuable contributions to improved diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunxi Huang
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University, Xi Guang, China
| | - Hong Huang
- Guilin Medical University, Xi Guang, China
| | - Zehua Wang
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China
| | - Xueyuan Jia
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Zhenjun Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruichong Lin
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China
- School of Computer Engineering, Guangzhou Huali College, Guangzhou, China
| | - Yue Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yisitandaer yalikun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Langping Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Li
- Burning Rock Biotech, Guangzhou, China
| | - Fei Zhao
- Burning Rock Biotech, Guangzhou, China
| | - Zhange Chen
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao, China
| | - Wenting Li
- Singleron Biotechnologies, Guangzhou, China
| | - Jianwei Liao
- Cellular and Molecular Diagnostics Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Miaoyun Long
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Thyroid Surgery, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Abel J, Jain S, Rajan D, Padigela H, Leidal K, Prakash A, Conway J, Nercessian M, Kirkup C, Javed SA, Biju R, Harguindeguy N, Shenker D, Indorf N, Sanghavi D, Egger R, Trotter B, Gerardin Y, Brosnan-Cashman JA, Dhoot A, Montalto MC, Parmar C, Wapinski I, Khosla A, Drage MG, Yu L, Taylor-Weiner A. AI powered quantification of nuclear morphology in cancers enables prediction of genome instability and prognosis. NPJ Precis Oncol 2024; 8:134. [PMID: 38898127 PMCID: PMC11187064 DOI: 10.1038/s41698-024-00623-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
While alterations in nucleus size, shape, and color are ubiquitous in cancer, comprehensive quantification of nuclear morphology across a whole-slide histologic image remains a challenge. Here, we describe the development of a pan-tissue, deep learning-based digital pathology pipeline for exhaustive nucleus detection, segmentation, and classification and the utility of this pipeline for nuclear morphologic biomarker discovery. Manually-collected nucleus annotations were used to train an object detection and segmentation model for identifying nuclei, which was deployed to segment nuclei in H&E-stained slides from the BRCA, LUAD, and PRAD TCGA cohorts. Interpretable features describing the shape, size, color, and texture of each nucleus were extracted from segmented nuclei and compared to measurements of genomic instability, gene expression, and prognosis. The nuclear segmentation and classification model trained herein performed comparably to previously reported models. Features extracted from the model revealed differences sufficient to distinguish between BRCA, LUAD, and PRAD. Furthermore, cancer cell nuclear area was associated with increased aneuploidy score and homologous recombination deficiency. In BRCA, increased fibroblast nuclear area was indicative of poor progression-free and overall survival and was associated with gene expression signatures related to extracellular matrix remodeling and anti-tumor immunity. Thus, we developed a powerful pan-tissue approach for nucleus segmentation and featurization, enabling the construction of predictive models and the identification of features linking nuclear morphology with clinically-relevant prognostic biomarkers across multiple cancer types.
Collapse
|
4
|
Meng Z, Wang G, Su F, Liu Y, Wang Y, Yang J, Luo J, Cao F, Zhen P, Huang B, Yin Y, Zhao Z, Guo L. A Deep Learning-Based System Trained for Gastrointestinal Stromal Tumor Screening Can Identify Multiple Types of Soft Tissue Tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:899-912. [PMID: 37068638 DOI: 10.1016/j.ajpath.2023.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
The accuracy and timeliness of the pathologic diagnosis of soft tissue tumors (STTs) critically affect treatment decision and patient prognosis. Thus, it is crucial to make a preliminary judgement on whether the tumor is benign or malignant with hematoxylin and eosin-stained images. A deep learning-based system, Soft Tissue Tumor Box (STT-BOX), is presented herein, with only hematoxylin and eosin images for malignant STT identification from benign STTs with histopathologic similarity. STT-BOX assumed gastrointestinal stromal tumor as a baseline for malignant STT evaluation, and distinguished gastrointestinal stromal tumor from leiomyoma and schwannoma with 100% area under the curve in patients from three hospitals, which achieved higher accuracy than the interpretation of experienced pathologists. Particularly, this system performed well on six common types of malignant STTs from The Cancer Genome Atlas data set, accurately highlighting the malignant mass lesion. STT-BOX was able to distinguish ovarian malignant sex-cord stromal tumors without any fine-tuning. This study included mesenchymal tumors that originated from the digestive system, bone and soft tissues, and reproductive system, where the high accuracy of migration verification may reveal the morphologic similarity of the nine types of malignant tumors. Further evaluation in a pan-STT setting would be potential and prospective, obviating the overuse of immunohistochemistry and molecular tests, and providing a practical basis for clinical treatment selection in a timely manner.
Collapse
Affiliation(s)
- Zhu Meng
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Guangxi Wang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fei Su
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Beijing Key Laboratory of Network System and Network Culture, Beijing, China
| | - Yan Liu
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuxiang Wang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jing Yang
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Panpan Zhen
- Department of Pathology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Binhua Huang
- Department of Pathology, Dongguan Houjie Hospital, Dongguan, China
| | - Yuxin Yin
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhicheng Zhao
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China; Beijing Key Laboratory of Network System and Network Culture, Beijing, China.
| | - Limei Guo
- Beijing University of Posts and Telecommunications and Department of Pathology, Peking University Third Hospital, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
5
|
Michielli N, Caputo A, Scotto M, Mogetta A, Pennisi OAM, Molinari F, Balmativola D, Bosco M, Gambella A, Metovic J, Tota D, Carpenito L, Gasparri P, Salvi M. Stain normalization in digital pathology: Clinical multi-center evaluation of image quality. J Pathol Inform 2022; 13:100145. [PMID: 36268060 PMCID: PMC9577129 DOI: 10.1016/j.jpi.2022.100145] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/20/2022] Open
Abstract
In digital pathology, the final appearance of digitized images is affected by several factors, resulting in stain color and intensity variation. Stain normalization is an innovative solution to overcome stain variability. However, the validation of color normalization tools has been assessed only from a quantitative perspective, through the computation of similarity metrics between the original and normalized images. To the best of our knowledge, no works investigate the impact of normalization on the pathologist's evaluation. The objective of this paper is to propose a multi-tissue (i.e., breast, colon, liver, lung, and prostate) and multi-center qualitative analysis of a stain normalization tool with the involvement of pathologists with different years of experience. Two qualitative studies were carried out for this purpose: (i) a first study focused on the analysis of the perceived image quality and absence of significant image artifacts after the normalization process; (ii) a second study focused on the clinical score of the normalized image with respect to the original one. The results of the first study prove the high quality of the normalized image with a low impact artifact generation, while the second study demonstrates the superiority of the normalized image with respect to the original one in clinical practice. The normalization process can help both to reduce variability due to tissue staining procedures and facilitate the pathologist in the histological examination. The experimental results obtained in this work are encouraging and can justify the use of a stain normalization tool in clinical routine.
Collapse
Affiliation(s)
- Nicola Michielli
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Alessandro Caputo
- Department of Medicine and Surgery, University Hospital of Salerno, Salerno, Italy
| | - Manuela Scotto
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Alessandro Mogetta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Orazio Antonino Maria Pennisi
- Technology Transfer and Industrial Liaison Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Filippo Molinari
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Davide Balmativola
- Pathology Unit, Humanitas Gradenigo Hospital, Corso Regina Margherita 8, 10153 Turin, Italy
| | - Martino Bosco
- Department of Pathology, Michele and Pietro Ferrero Hospital, 12060 Verduno, Italy
| | - Alessandro Gambella
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Jasna Metovic
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Daniele Tota
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Laura Carpenito
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- University of Milan, Milan, Italy
| | - Paolo Gasparri
- UOC di Anatomia Patologica, ASP Catania P.O. “Gravina”, Caltagirone, Italy
| | - Massimo Salvi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| |
Collapse
|